• Aucun résultat trouvé

Stress responses, outer membrane permeability control and antimicrobial resistance in Enterobacteriaceae 2 3

N/A
N/A
Protected

Academic year: 2021

Partager "Stress responses, outer membrane permeability control and antimicrobial resistance in Enterobacteriaceae 2 3"

Copied!
24
0
0

Texte intégral

(1)

HAL Id: hal-01840466

https://hal.archives-ouvertes.fr/hal-01840466

Submitted on 16 Jul 2018

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

Stress responses, outer membrane permeability control and antimicrobial resistance in Enterobacteriaceae 2 3

Sushovan Dam, Jean-Marie Pages, Muriel Masi

To cite this version:

Sushovan Dam, Jean-Marie Pages, Muriel Masi. Stress responses, outer membrane permeability con- trol and antimicrobial resistance in Enterobacteriaceae 2 3. Microbiology, Microbiology Society, 2018,

�10.1099/mic.0.000613�. �hal-01840466�

(2)

Stress responses, outer membrane permeability control and antimicrobial resistance in

1  

Enterobacteriaceae

2  

3  

Sushovan Dam, Jean-Marie Pagès* and Muriel Masi

4  

5   6   7  

UMR_MD-1, Aix-Marseille Univ. & IRBA, 27 Boulevard Jean Moulin, 13005 Marseille,

8  

France.

9   10  

*Corresponding author:

11  

jean-marie.pages@univ-amu.fr 12  

+33 (0)4 91 32 46 97

13  

14  

Key words: Enterobacteriaceae, envelope stress responses, outer membrane permeability,

15  

porins, drug translocation, multidrug resistance.

16   17  

Category: Regulation

18  

19  

Word count (from introduction to conclusion):

20   21  

Abbreviations: outer membrane (OM), inner membrane (IMI), peptidoglycan (PG),

22  

lipopolysaccharide (LPS), antimicrobial resistance (AMR), multidrug resistance (MDR),

23  

envelope stress response (ESR), two-component system (TCS), small regulatory RNA

24  

(sRNA).

25   26  

(3)

Abstract

27  

Bacteria have evolved several strategies to survive a myriad of harmful conditions in the

28  

environment and in hosts. In Gram-negative bacteria, responses to nutrient limitation,

29  

oxidative or nitrosative stress, envelope stress, exposure to antimicrobials and other growth-

30  

limiting stresses have been linked to the development of antimicrobial resistance. This results

31  

from the activation of protective changes to cell physiology (decreased outer membrane

32  

permeability), resistance transporters (drug efflux pumps), resistant lifestyles (biofilms,

33  

persistence) and/or resistance mutations (target mutations, production of antibiotic

34  

modification/degradation enzymes). In targeting and interfering with essential physiological

35  

mechanisms, antimicrobials themselves are considered as stresses to which protective

36  

responses have also evolved. In this review, we focus on envelope stress responses that affect

37  

the expression of outer membrane porins and their impact on antimicrobial resistance. We

38  

also discuss evidences that indicate the role of antimicrobials as signaling molecules in

39  

activating envelope stress responses.

40  

(4)

Introduction

41  

Antimicrobial resistance (AMR) is broadly recognized as a growing threat for human health

42  

[1, 2, 3]. As such, increasing antibiotic treatment failures due to multidrug resistant (MDR)

43  

bacteria have stirred the urgent need to better understand the underlying molecular

44  

mechanisms and promote innovation with the development of new antibiotics and alternative

45  

therapies [4, 5]. The efficacy of antibacterial compounds depends on their capacity to reach

46  

inhibitory concentrations at the vicinity of their target. This is particularly challenging for

47  

drugs directed against Gram-negative bacteria, which exhibit a complex envelope comprising

48  

two membranes and transmembrane efflux pumps [6, 7]. The Gram-negative envelope

49  

comprises an inner membrane (IM), which is a symmetric phospholipid bilayer; a thin

50  

peptidoglycan (PG) layer ensuring the cell shape; and an outer membrane (OM) that is an

51  

asymmetric bilayer, composed of an inner leaflet of phospholipids and an outer leaflet of

52  

lipopolysaccharide (LPS) [8]. The OM is a barrier to both hydrophobic and hydrophilic

53  

compounds, including necessary nutrients, metabolic substrates and antimicrobials, but

54  

access is provided by the water filled

β-barrel channels called porins [6, 9, 10, 11, 12]. In 55  

Escherichia coli, the channels of the general porins OmpF and OmpC are size restricted and

56  

show a preference for passage of hydrophilic charged compounds, including antibiotics such

57  

as

β-lactams and fluoroquinolones. These porins are conserved throughout the phylum of γ

-

58  

proteobacteria [13]. Additionally, tripartite RND (Resistance-Nodulation-cell Division)

59  

efflux pumps, such as AcrAB-TolC in E. coli, play a major role in removing antibiotics from

60  

the periplasm [7, 12]. Not surprisingly, MDR clinical isolates of Enterobacteriaceae

61  

generally exhibit porin loss and/or increased efflux, which act in synergy to reduce the

62  

intracellular accumulation of antibiotics below the threshold that would be efficient for

63  

activity [10].

64  

Given the importance of the OM in controlling the uptake of beneficial as well as toxic

65  

compounds, one can expect that the expression of porins depends on environmental stresses

66  

and is well-coordinated at the transcriptional and post-transcriptional levels [10, 14-17]. In

67  

this review, we will address the porin-mediated influx of antibiotics and give a perspective on

68  

the factors, including major regulatory pathways and antibiotic stresses, which control porin

69  

expression in E. coli and closely relative Enterobacteriaceae. Additionally, we will discuss

70  

the recent clinical data that illustrate the bacterial strategies using porins modifications to

71  

limit antibiotic entry.

72   73  

(5)

Antibiotic stresses

74  

Bacteria are present in a wide range of environments in which they are exposed to diverse

75  

toxic compounds or growth-limiting conditions. These include antibiotics used in the medical

76  

environment and agricultural settings. The last few decades have been marked by the constant

77  

increase of (multi)drug resistant clinical isolates to which we responded by increasing

78  

antibiotic dosing. Therefore, antibiotics are present almost everywhere at different

79  

concentrations [18]. Although MDR still emerges from bacterial exposure to antibiotic

80  

concentrations that are higher than the minimal inhibitory concentrations (MIC, defined as

81  

the lowest concentration of a drug that inhibit bacterial growth in defined laboratory

82  

conditions), the effects of subinhibitory concentrations on bacterial physiology and AMR was

83  

mostly disregarded. Importantly, studies in this field have shown that low antibiotic

84  

concentrations affect bacteria at least at four different levels: as selectors of resistance (by

85  

enriching resistant bacteria within populations and selecting for de novo resistance mutations)

86  

[19]; (ii) as contributors of genetic and phenotypic heterogeneity [20]; (iii) as intercellular

87  

signals [21]; (iv) as inducers of persistence [22]. In this regard, Viveiros and colleagues have

88  

demonstrated the induction of high-level resistance to tetracycline (TET) in susceptible E.

89  

coli K12 obtained by gradual, step-wise increase exposure to subinhibitory concentrations of

90  

the antibiotic [23]. Increased expression of the AcrAB efflux pump was found responsible for

91  

resistance to TET, which could also be reversed by the use of the efflux pump inhibitor

92  

phenylalanine-arginine-β-naphthylamide (PAβN). Interestingly, the TET-resistant strain also

93  

exhibited MDR due to repression of OmpF and OmpC expression [24]. Important questions

94  

arise from this and other related studies. First is whether the target for signaling resistance is

95  

the same as the target that is inhibited by the antibiotic. In case the antibiotic itself but not a

96  

secondary metabolite is the signaling molecule, this could be determined by examining

97  

whether the response is alleviated by a target mutation that prevents drug binding. Second is

98  

whether and how the antibiotic (or a secondary metabolite) interferes with the ESRs

99  

described above. Here, comparative transcriptomics between susceptible and resistant strains

100  

would be a valuable tool to answer this question.

101   102  

Global regulators

103  

In Enterobacteriaceae, the development of MDR is under positive regulation by global

104  

transcriptional activators that include members of Ara/XylS superfamily such as MarA,

105  

RamA (absent in E. coli) and Rob as well as the oxidative stress regulon SoxSR [10, 25-29].

106  

Mutations in the corresponding genes are well-documented and induce the overproduction of

107  

(6)

efflux pumps with concomitant repression of porin expression both directly and indirectly

108  

[10]. These mechanisms are reviewed in details in Davin et al. [10]. Negative regulation by

109  

repressors of porins also plays a major role. OmpX is a small OM channel [30], of which

110  

overexpression is associated with a decreased expression of Omp36 (the OmpC ortholog of

111  

Enterobacter aerogenes) and a decreased susceptibility to β-lactams [31, 32]. Studies have

112  

indicated that expression OmpX itself is controlled by a number of environmental factors

113  

including salicylate via MarA and paraquat via SoxS [33] A very rapid MarA-dependent

114  

response pathway for upregulation of ompX has been shown to occur within 60-120 min upon

115  

cell exposure to salicylate [32]. This work by Dupont et al. identified dramatic decrease in

116  

OmpF levels, as a first line of defense, with simultaneous development of resistance to

β- 117  

lactams and fluoroquinolones by altering OM permeability.

118   119  

Envelope stress responses

120  

All living organisms have stress responses that allow them to sense and respond to

121  

environmental damaging conditions by remodeling gene expression. As such, Gram-negative

122  

bacteria possess stress responses that are uniquely targeted to the cell envelope, including

123  

membranes and cell wall. These envelope stress responses (ESRs) are the EnvZ/OmpR,

124  

CpxAR (Cpx), BaeRS, and Rcs phosphorelays, the stress responsive alternative sigma factor

125  

σE

, and the phage shock response [34-37] in E. coli and closely related Enterobacteriaceae.

126  

Each of these ESRs is activated following the perturbation of particular components of the

127  

envelope or exposure to particular environmental stresses. Although ESRs are important for

128  

reacting to damaging conditions, stress proteins also play important roles in the maintenance

129  

of basic cellular physiology [38, 39]. This is particularly true for the

σE

-dependent stress

130  

response in E. coli, as the rpoE gene, which encodes σ

E

, is essential for viability [40]. Here,

131  

we will essentially focus on ESRs that impact on AMR by regulating porin expression

132  

together with many other targets (regulons) — namely EnvZ/OmpR, Cpx and σ

E

(see below

133  

and key figure). Additionally, with the recent highlights and advances in RNA-based

134  

techniques [41], the repertoire of small regulatory RNAs (sRNAs) has vastly increased so as

135  

to and their impact on the OM is continuously emerging [15, 17]. sRNAs alter gene

136  

expression, allowing fast adjustment to different growth conditions [42]. Noteworthy, ESRs

137  

are often interconnected, regulate and are regulated by sRNAs in order control target genes

138  

both at the transcriptional and post-transcriptional levels [15-17, 43, 44] (see below and key

139  

figure).

140  

(7)

Osmolarity was one of the earliest stresses described to influence OmpF and OmpC

141  

expression via the EnvZ/OmpR two-component system (TCS) [45, 46]. EnvZ is a membrane-

142  

bound sensor kinase, and OmpR is a cytosolic response regulator, which binds to the

143  

promoter region of the porin genes. Upon activation, EnvZ autophosphorylates and the high

144  

energy phosphoryl group from EnvZ is subsequently transferred to a conserved Asp residue

145  

on OmpR. Phosphorylated OmpR (OmpR~P) serves as a transcription factor that

146  

differentially modulates the expression of the ompF and ompC porin genes [45]. At low

147  

osmolarity, high levels of OmpR~P activates ompF transcription, whereas at high osmolarity,

148  

low levels of OmpR~P represses ompF transcription and activates ompC transcription [47].

149  

This differential production of OmpF and OmpC is consistent with that in high osmolarity

150  

environments, such as in the hosts where nutrients are abundant the least permeable pore

151  

channel OmpC is predominant, thus limiting the uptake of toxic bile salts; whereas in low

152  

osmolarity environments where nutrients are scarce, the most permeable pore channel OmpF

153  

is expressed [6]. OmpF and OmpC transcriptional regulation by EnvZ/OmpR is also triggered

154  

by local anesthetics, pH, and nutrient limitation [46].

155  

Accumulation of misfolded OM proteins in the periplasm, presumably reflecting problems in

156  

protein assembly or transport across the IM, can be detected by regulatory sensors that

157  

activate either the Cpx TCS or the alternative sigma factor

σE

.

σE

and Cpx are the two major

158  

regulation pathways that control the envelop integrity with overlapping regulon members

159  

[48-51] but respond to different inducing cues [35]. It is possible that these poorly defined

160  

signals (see below) act by causing accumulation of misfolded proteins. However, misfolded

161  

proteins are not the inducing signal per se, as some induce

σE

but not Cpx and vice versa.

162  

Recent studies rather suggest that Cpx responds to IM perturbations, while σ

E

is activated by

163  

signals at the OM. The Cpx system comprises the CpxA sensor kinase and response regulator

164  

CpxR. Envelope stresses including alkaline pH, periplasmic protein misfolding, IM

165  

abnormalities such as misfolded transporters or accumulation of the lipid II precursor, induce

166  

the dissociation of the accessory protein CpxP from CpxA, trigger CpxA-mediated

167  

phosphorylation of CpxR, and altered expression of protein foldases and proteases,

168  

respiratory complexes, IM transporters, and cell wall biogenesis enzymes [37, 48, 49], all of

169  

which affect resistance to a number of antibiotics, particularly aminoglycosides and

β- 170  

lactams [37, 49, 52-54]. The Cpx-mediated regulation of porins occurs at several levels. At

171  

the transcriptional level, CpxR~P has been shown to bind directly the ompF and ompC

172  

promoters [55]. More recently, it has been found that the small IM protein MzrA connects

173  

(8)

Cpx and EnvZ/OmpR [56]. In this pathway and upon the activation of Cpx, MzrA interacts

174  

directly with EnvZ, which in turn, stabilizes OmpR~P [57]. In sensing different signals, the

175  

interconnection between Cpx and EnvZ/OmpR allows cells to adapt to diverse environmental

176  

stresses. Finally, although Cpx contributes to AMR by regulating a number of genes [37, 49,

177  

52-54], its precise role and that of other TCSs in the development of MDR in clinical isolates

178  

are still poorly documented [58]. On the other hand, the stress responsive sigma factor σ

E

is

179  

induced by stresses that disturb the OM and its regulon members comprise genes that

180  

facilitate the biogenesis of OM components, including proteins, lipoproteins and LPS [59-

181  

67]. In the absence of inducing signals,

σE

is held at the cytoplasmic side of the IM by the

182  

anti-sigma factor RseA. At the periplasmic side of the IM, RseB binds to RseA, thus

183  

enhancing the inhibition of

σE

. Upon activation,

σE

is released from RseA by a proteolytic

184  

cascade that starts with the sequential degradation of the periplasmic and transmembrane

185  

domains of RseA by DegS and RseP, respectively, followed by the degradation of the

186  

cytoplasmic domain of RseA by ClpXP [68]. Interestingly, proteolysis of RseA is triggered

187  

by the binding of a conserved peptide found at the C-terminus of OM proteins, which is

188  

normally buried in folded porin trimers, to DegS in conjunction with the release of RseB

189  

from RseA upon binding of LPS intermediates [69, 70]. Of note, the σ

E

-dependent repression

190  

of porin synthesis only occurs at the post-transcriptional level, wherein base-paring sRNAs

191  

inhibits translation of omp mRNAs (see below) in order to maintain the envelope homeostasis

192  

under stress conditions, as porins are major abundant proteins under normal growth

193  

conditions [6].

194  

The post-transcriptional repression of OmpF by the sRNA MicF has been discovered in 1984

195  

[71-73]. This 93-nucleotide (nt) RNA is transcribed in the opposite direction to the ompC

196  

gene and acts by direct base-pairing to a region that encompasses the ribosome binding site

197  

(RBS) and the start codon of the ompF mRNA, thus preventing translation initiation [74].

198  

The expression of the MicF sRNA itself is subjected to multiple signals and regulatory

199  

pathways [75]. Positive regulation includes EnvZ/OmpR in high osmolarity conditions [76],

200  

SoxS in response to oxidative stress [77] and MarA in response to antibiotic stress [25]. The

201  

109-nt MicC sRNA has been identified more recently and shown to repress OmpC by direct

202  

base-pairing to a 5’ untranslated region of the ompC mRNA [78]. Interestingly, micC is

203  

transcribed in the opposite direction to the ompN gene that encodes a quiescent porin

204  

homologous to OmpF and OmpC [79]. We have recently shown that ompN and micC are

205  

subjected to dual regulation upon exposure to certain antimicrobials such as

β-lactams in a 206  

(9)

σE

-dependent manner [80]. This is consistent with that ompN-micC and ompC-micF share

207  

similar genetic organization and that ompC and micF are co-induced under specific

208  

conditions (i. e. high osmolarity via EnvZ/OmpR). The last decade has been marked by the

209  

identification and characterization of several sRNAs. These are differentially expressed and

210  

have been assigned to various important regulatory pathways in E. coli and Salmonella.

211  

Interestingly, many pathways regulate and are regulated by sRNAs [43, 44]. A prime

212  

example is EnvZ/OmpR, which activates the expression of MicF (that target ompF), OmrA

213  

and OmrB (that target ompT and mRNA of OM channels for iron-siderophore complexes)

214  

[81]; OmrA and OmrB, in turn, repress the translation of the ompR mRNA, creating a

215  

negative feedback loop [82]. Others examples include the well-conserved

σE

-regulated

216  

sRNAs RybB (that target ompC and lamB in E. coli; ompN and ompW in Salmonella), MicA

217  

(ompA), RseX (ompC and ompA), CyaR (ompX) and MicL (that represses translation of the

218  

major OM lipoprotein Lpp) [43, 66, 83-90] (key figure). Of note all these sRNAs are trans-

219  

acting, function by imperfect base pairing with multiple mRNA targets and require the help

220  

of the RNA chaperone Hfq [15-17].

221   222  

Porin alterations in clinical isolates

223  

Combined regulations contributed by different stressors leads to hampering of the drug

224  

accumulation inside cells under the threshold for bacterial death. In one such study in K.

225  

pneumoniae, preferential expression of OmpK37 was detected in porin-deficient strains [92].

226  

Amino acid sequencing showed that OmpK37 is highly homologous to quiescent porins

227  

OmpS2 from Salmonella enterica serovar Typhimurium and OmpN from E. coli. Liposome

228  

swelling assay with purified porins determined that OmpK37 also has a narrower pore, which

229  

was responsible for higher MICs of cefotaxime and cefoxitin antibiotics because of lower

230  

drug diffusion. A very recent study identified mutation in the pho regulon of an extensively

231  

drug resistant strain of K. pneumoniae demonstrating downregulation of phoE gene by

232  

mutations in phoR and phoB. Here the PhoE porin, which is normally involved in phosphate

233  

transport, promotes restoration of cefoxitin and carbapenem resistance [93]. This is an

234  

interesting example of a regulatory mutation that effects porin expression, and clinically

235  

favors AMR under antibiotic therapy.

236  

A wide array of chemicals including disinfectants and antibiotics has been shown to modulate

237  

the OM permeability including expression of porins [94]. In addition, several studies have

238  

(10)

described the effect of imipenem on porin loss or loss of function mutations in clinical

239  

isolates of Enterobacteriaceae [58, 95-100].

240  

Porins are trimers of 16-stranded

β-barrels, each monomer formed of a central channel 241  

constricted by loop 3 that folds inward, thereby restricting the size of the channel. The

242  

presence of acidic residues in loop 3 facing a cluster of basic residues on the opposite side of

243  

the pore creates a strong transversal electric field [6, 101, 102]. This so-called eyelet or

244  

constriction region determines the channel size and ion selectivity, with OmpF being more

245  

permeable than OmpC. This latter observation was first attributed to the OmpC pore being

246  

slightly more constricted in this porin compared to OmpF [101, 102]. Although the two

247  

porins share high sequence similarity, the pore interior is more negative in OmpC than in

248  

OmpF [102]. This can also account for the low permeability of OmpC for anionic β-lactams

249  

[103, 104]. Moreover, the replacement of all ten titratable residues that differ between OmpC

250  

and OmpF in the pore-lining region leads to the exchange of antibiotic permeation properties

251  

[105]. Together, these structural and functional data clearly demonstrate that the charge

252  

distribution at pore linings, but not pore size, is a critical parameter that physiologically

253  

distinguishes OmpC from OmpF.

254  

Functional mutations in porin genes leading to reduced permeability are another strategy

255  

found in MDR bacteria. In two documented cases,

β-lactam-resistant clinical isolates of

E.

256  

aerogenes contained Omp36 (an OmpC homologue) that carried the mutation G112D in L3

257  

[106, 97]. The homologous mutation G119D in OmpF of E. coli narrows the size of the

258  

channel as the large side chain of Asp protrudes into the channel lumen and confers a drastic

259  

reduction in

β-lactam susceptibility [107]. Consistently, the Omp36 G112D mutant of

E.

260  

aerogenes was characterized by a 3-fold decrease in ion conductance and a significant

261  

decrease in cephalosporin sensitivity (e. g. MICs of cefotaxime, cefpirome, cefepime and

262  

ceftazidime were 7 to 9 fold higher in the clinical isolate as compared to that in a sensitive

263  

reference strain) and a cross resistance to carbapenems [106, 97]. Recent studies also found a

264  

series of OmpC mutants that were isolated from a patient with chronic E. coli infection and

265  

additive mutations that conferred increased resistance to a variety of antibiotics, including

266  

cefotaxime, ceftazidime, imipenem, meropenem and ciprofloxacin [108, 109].

 

Low et al.

267  

demonstrated that subtle changes in OmpC in clinical isolates of E. coli altered antibiotic

268  

permeability and thus cell viability [108]. Seven isolates collected over a two year clinical

269  

treatment exhibited increased levels of antibiotic resistance. These isolates exhibited the same

270  

two mutations (D18E and S274F) in the OmpC porin with increased levels of antibiotic

271  

(11)

resistance, thus pointing towards the possible functional role of these mutations in antibiotic

272  

influx.

273  

It is worthwhile to note that from our knowledge, porin mutations causing reduced

274  

permeability have only been described in OmpC-type porins in E coli and E aerogenes.

275  

Interestingly, this type of porin is expressed under high osmolarity, the same environment the

276  

bacteria encounters the hosts. This gives an essential outlook on the host induced

277  

modifications that possibly occurs in these pathogens during infection. Heeding to this sort of

278  

information can be highly beneficial for designing drugs with an improved diffusion across

279  

the bacterial outer membrane.

280   281  

Conclusion

282  

It is noteworthy that the sRNA-mediated stress response mechanism has multiple benefits for

283  

bacteria as compared to regulation by protein. Since sRNAs are produced during

284  

transcription, the later stages of translation and post translational modification processes in

285  

the cell is completely skipped proving to be time and energy efficient for the cell. Not to

286  

forget the energy saved in porin assembly and discarding of misfolded proteins, which in

287  

itself can induce another stress response mechanism.

288  

Decreased porin expression has been observed as a rapid response to toxic molecules and

289  

antibiotics within less than 60 minutes. Many sRNAs act at the post-transcriptional level,

290  

which ensures a rapid response to stressful conditions. In addition, the versatility of sRNAs

291  

ensures another level of gene regulation along with protein transcriptional regulators, thus

292  

contributing to an additional layer of tighter regulation. Taking into account the major role of

293  

the CpxAR and EnvZ/OmpR regulators in response to stressors such as antibiotics, it will be

294  

interesting to develop some assays allowing the detection of these kinds of mutations inside

295  

clinical isolate. This original diagnostic maybe used for determining the prevalence of these

296  

regulation events in clinical strain that have undergone antibiotic stress.

297  

Targeting the early transcriptional step of antibiotic stress response regulatory mechanism is

298  

much more logical, especially when we have reports of OMP expression being regulated

299  

(both up and downregulation) within 60 minutes of stress appearance [32]. This will

300  

especially promote bypassing of aforementioned mutations in porins in clinical strains that

301  

are selected during antibiotic treatment. Targeting of sRNA or sRNA regulators such as

302  

MicF or Hfq can rejuvenate failing antimicrobial therapies in regards with membrane

303  

impermeability. They can be original targets for increasing the efficiency of existing drugs by

304  

providing fitness reduction in bacteria. As of now, a cyclic peptide RI20 has been identified

305  

(12)

to inhibit Hfq-mediated repression of gene, by binding with proximal binding site of Hfq

306  

[110]. Another approach will be to inhibit sRNA interfering with porin expression that is

307  

involved in drug translocation. Recently, a small molecule was used to target human

308  

microRNA (miR)-525 precursors as an anti-cancer strategy [111]. This promising discovery

309  

can be repeated in bacteria for manipulating sRNA levels, which may save the failing

310  

antibiotic therapies.

311  

Predictability of an efficient drug based on the SICAR (Structure Intracellular Concentration

312  

Activity Relationship) concept, is a step up to efficient drug designing. Briefly, SICAR

313  

connects the physicochemical drug properties to the efficacy of translocation through the

314  

bacterial membrane and the resulting intracellular accumulation. To achieve this goal, an

315  

extensive knowledge of the OM permeability control, including the contribution of sRNAs, is

316  

required.

317  

(13)

Funding information: The research leading to the discussions presented here was conducted

318  

as part of the Marie Curie Initial Training Network TRANSLOCATION consortium and has

319  

received support from the ITN-2013-607694-Translocation (SD). This work was also

320  

supported by Aix-Marseille Univ. and Service de Santé des Armées.

321   322  

Acknowledgments: We thank all the members of the UMR_MD1, especially Estelle Dumont

323  

and Julia Vergalli, for helpful discussions throughout this work.

324   325  

Conflicts of interest: The authors declare no conflict of interest. The founding sponsors had

326  

no role in the design of the study; in the collection, analyses, or interpretation of data; in the

327  

writing of the manuscript, and in the decision to publish the results.

328  

(14)

References

329  

1. O’Neill, J. http://amr-review.org/sites/default/files/Tackling%20drug-resistant%

330  

20infections%20%20An%20overview%20of%20our%20work_IncVaccineLR_NOCROPS.p

331  

df (accessed March 3, 2016).

332  

2. WHO. http://apps.who.int/iris/bitstream/10665/112642/1/9789241564748_eng.pdf

333  

(accessed March 3, 2016).

334  

3. NIAID. http://www.niaid.nih.gov/topics/antimicro

335  

topics/antimicrobialresistance/documents/arstrategicplan2014.pdf (accessed March 3, 2016).

336  

4. Stavenger RA, Winterhalter M. TRANSLOCATION project: how to get good

337  

drugs into bad bugs. Sci Transl Med 2014;6:228ed7.

338  

5. Laxminarayan R, Matsoso P, Pant S, Brower C, Røttingen, JA

et al. Access to 339  

effective antimicrobials: a worldwide challenge. The Lancet 2016;384:168-175.

340  

6. Nikaido H. Molecular basis of bacterial outer membrane permeability revisited.

341  

Microbiol Mol Biol Rev 2003;67:593-656.

342  

7. Nikaido H, Pagès JM. Broad-specificity efflux pumps and their role in multidrug

343  

resistance of Gram-negative bacteria. FEMS Microbiol Rev 2012;36:340-63.

344  

8. Silhavy TJ, Kahne D, Walker S. The bacterial cell envelope. Cold Spring Harb

345  

Perspect Bio. 2010;2:a000414.

346  

9. Pagès JM, James, CE, Winterhalter M. The porin and the permeating antibiotic: a

347  

selective diffusion barrier in Gram-negative bacteria. Nat Rev Microbiol 2008;6:893-903.

348  

10. Davin-Régli A, Bolla JM, James CE, Lavigne JP, Chevalier J

et al.

Membrane

349  

permeability and regulation of drug "influx and efflux" in enterobacterial pathogens. Curr

350  

Drug Targets 2008;9:750-9.

351  

11. Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim

352  

Biophys Acta 2009;1794:808-16.

353  

12. Masi M, Réfrégiers M, Pos KM, Pagès JM. Mechanisms of envelope permeability

354  

and antibiotic influx and efflux in Gram-negative bacteria. Nat Microbiol 2017;2:17001.

355  

13. Nguyen TX, Alegre ER, Kelley ST. Phylogenetic analysis of general bacterial

356  

porins: a phylogenomic case study. J Mol Microbiol Biotechnol 2006;11:291-301.

357  

14. Masi M, Pagès JM. Structure, function and regulation of outer membrane proteins

358  

involved in drug transport in Enterobactericeae: the OmpF/C-TolC Case. Open Microbiol J

359  

2013;7:22–33.

360  

15. Guillier M, Gottesman S, Storz G. Modulating the outer membrane with small

361  

(15)

16. Vogel J, Papenfort K. Small non-coding RNAs and the bacterial outer membrane.

363  

Curr Opin Microbiol 2006;9:605-611.

364  

17. Valentin-Hansen P, Johansen J, Rasmussen AA. Small RNAs controlling outer

365  

membrane porins. Curr Opin Microbiol 2007;10(2):152-155.

366  

18. Andersson DI, Hughes D. Microbiological effects of sublethal levels of antibiotics.

367  

Nat Rev Microbiol 2014;12:465-478.

368  

19. Gullberg E, Cao S, Berg OG, Ilbäck C, Sandegren L

et al. Selection of resistant 369  

bacteria at very low antibiotic concentrations. PLoS Pathog 2011;7:e1002158.

370  

20. Gutierrez A, Laureti L, Crussard S, Abida H, Rodríguez-Rojas A et al. β-lactam

371  

antibiotics promote bacterial mutagenesis via an RpoS-mediated reduction in replication

372  

fidelity. Nat Commun 2013;4:1610.

373  

21. Lee HH, Molla MN, Cantor CR, Collins JJ. Bacterial charity work leads to

374  

population-wide resistance. Nature 2010;467:82-85.

375  

22. Pu Y, Zhao Z, Li Y, Zou J, Ma Q

et al. Enhanced efflux activity facilitates drug 376  

tolerance in dormant bacterial cells. Mol Cell 2016;62:284-294.

377  

23. Viveiros M, Jesus A, Brito M, Leandro C, Martins M

et al. Inducement and 378  

reversal of tetracycline resistance in Escherichia coli K-12 and expression of proton gradient-

379  

dependent multidrug efflux pump genes. Antimicrob Agents Chemother 2005;49:3578-3582.

380  

24. Viveiros M, Dupont M, Rodrigues L, Couto I, Davin-Regli A

et al. Antibiotic 381  

stress, genetic response and altered permeability of E. coli. PLoS One 2007;2(4):e365.

382  

25. Cohen SP, McMurry LM, Levy SB. MarA locus causes decreased expression of

383  

OmpF porin in multiple-antibiotic-resistant (Mar) mutants of Escherichia coli. J Bacteriol

384  

1988;170:5416-5422.

385  

26. George AM, Hall RM, Stokes HW. Multidrug resistance in Klebsiella pneumoniae:

386  

a novel gene, ramA, confers a multidrug resistance phenotype in Escherichia coli.

387  

Microbiology 1995;141:1909-1920

388  

27. Chubiz LM, Rao CV. Role of the mar-sox-rob regulon in regulating outer membrane

389  

porin expression. J Bacteriol. 2011;193:2252-2260.

390  

28. Chollet R, Bollet C, Chevalier J, Malléa M, Pagès JM et al. mar operon involved

391  

in multidrug resistance of Enterobacter aerogenes. Antimicrob Agents Chemother

392  

2002;46:1093-1097.

393  

29. Chollet R, Chevalier J, Bollet C, Pagès JM, Davin-Régli A. RamA is an alternate

394  

activator of the multidrug resistance cascade in Enterobacter aerogenes. Antimicrob Agents

395  

Chemother 2004;48:2518-2523.

396  

(16)

30. Dupont M, Dé E, Chollet R, Chevalier J, Pagès JM. Enterobacter aerogenes

397  

OmpX, a cation-selective channel mar- and osmo-regulated. FEBS Lett 2004;569:27-30.

398  

31. Gayet S, Chollet R, Molle G, Pagès JM, Chevalier J. Modification of outer

399  

membrane protein profile and evidence suggesting an active drug pump in Enterobacter

400  

aerogenes clinical strains. Antimicrob Agents Chemother. 2003;47:1555-1559.

401  

32. Dupont M, James CE, Chevalier J, Pagès JM. An early response to environmental

402  

stress involves regulation of OmpX and OmpF, two enterobacterial outer membrane pore-

403  

forming proteins. Antimicrob Agents Chemother 2007;51:3190-3198.

404  

33. Mecsas J, Welch R, Erickson JW, Gross CA. Identification and characterization of

405  

an outer membrane protein, OmpX, in Escherichia coli that is homologous to a family of

406  

outer membrane proteins including Ail of Yersinia enterocolitica. J Bacteriol 1995;177:799-

407  

804.

408  

34. Majdalani N, Gottesman S. The Rcs phosphorelay: a complex signal transduction

409  

system. Annu Rev Microbiol 2005;59:379–405.

410  

35. Ruiz N, Silhavy TJ. Sensing external stress: watchdogs of the Escherichia coli cell

411  

envelope. Curr Opin Microbiol 2005;8:122-126.

412  

36. Rowley G, Spector M, Kormanec J, Roberts M. Pushing the envelope:

413  

extracytoplasmic stress responses in bacterial pathogens. Nat Rev Microbiol 2006;4:383-394.

414  

37. Raivio TL. Everything old is new again: an update on current research on the Cpx

415  

envelope stress response. Biochim Biophys Acta 2014;1843:1529-1541.

416  

38. Hayden JD, Ades SE. The extracytoplasmic stress factor, sigmaE, is required to

417  

maintain cell envelope integrity in Escherichia coli. PLoS One 2008;3(2):e1573.

418  

39. Delhaye A, Collet JF, Laloux G. Fine-Tuning of the Cpx envelope stress response is

419  

required for cell wall homeostasis in Escherichia coli. MBio 2016;7(1):e00047-16.

420  

40. De Las Penas A, Connolly L, Gross CA. SigmaE is an essential sigma factor in

421  

Escherichia coli. J Bacteriol 1997;179:6862-6864.

422  

41. Wassarman KM, Kiley PJ. Global approaches for finding small RNA and small

423  

open reading frame functions. J Bacteriol 2010;192:26-28.

424  

42. Gottesman S. The small RNA regulators of Escherichia coli: roles and mechanisms.

425  

Annu Rev Microbiol 2004;58:303-328.

426  

43. Thompson KM, Rhodius VA, Gottesman S. SigmaE regulates and is regulated by a

427  

small RNA in Escherichia coli. J Bacteriol 2007;189:4243-4256.

428  

44. Vogt SL, Evans AD, Guest RL, Raivio TL. The Cpx envelope stress response

429  

(17)

45. Mizuno T, Mizushima S. Signal transduction and gene regulation through the

431  

phosphorylation of two regulatory components: the molecular basis for the osmotic

432  

regulation of the porin genes. Mol Microbiol 1990;4:1077-1082.

433  

46. Pratt LA, Hsing W, Gibson KE, Silhavy TJ. From acids to osmZ: multiple factors

434  

influence synthesis of the OmpF and OmpC porins in Escherichia coli. Mol Microbiol

435  

1996;20:911-7.

436  

47. Cai SJ, Inouye M. EnvZ-OmpR interaction and osmoregulation in Escherichia coli. J

437  

Biol Chem 2002;277:24155-61.

438  

48. Price NL, Raivio TL. Characterization of the Cpx regulon in Escherichia coli strain

439  

MC4100. J Bacteriol 2009;191:1798-1815.

440  

49. Raivio TL, Leblanc SK, Price NL. The Escherichia coli Cpx envelope stress

441  

response regulates genes of diverse function that impact antibiotic resistance and membrane

442  

integrity. J Bacteriol 2013;195:2755-2767.

443  

50. Guest RL, Wang J, Wong JL, Raivio TL. A bacterial stress response regulates

444  

respiratory protein complexes to control envelope stress adaptation. J Bacteriol 2017;199:pii:

445  

e00153-17.

446  

51. Dartigalongue C, Missiakas D, Raina S. Characterization of the Escherichia coli

447  

sigma E regulon. J Biol Chem 2000;276:20866-20875.

448  

52. Guest RL, Raivio TL. Role of the Gram-negative envelope stress response in the

449  

presence of antimicrobial agents. Trends Microbiol 2016;24:377-390.

450  

53. Mahoney TF, Silhavy TJ. The Cpx stress response confers resistance to some, but

451  

not all, bactericidal antibiotics. J Bacteriol 2013;195(9):1869-74.

452  

54. Moreau PL. Protective role of the RpoE (σE) and Cpx envelope stress responses

453  

against gentamicin killing of nongrowing Escherichia coli incubated under aerobic,

454  

phosphate starvation conditions. FEMS Microbiol Lett 2014;357:151-156.

455  

55. Batchelor E, Walthers D, Kenney LJ, Goulian M. The Escherichia coli CpxA-

456  

CpxR envelope stress response system regulates expression of the porins OmpF and OmpC. J

457  

Bacteriol 2005;187:5723-5731.

458  

56. Gerken H, Charlson ES, Cicirelli EM, Kenney LJ, Misra R. MzrA: a novel

459  

modulator of the EnvZ/OmpR two-component regulon. Mol Microbiol. 2009;72:1408-1422.

460  

57. Gerken H, Misra R. MzrA-EnvZ interactions in the periplasm influence the

461  

EnvZ/OmpR two-component regulon. J Bacteriol 2010;192:6271-8.

462  

(18)

58. Philippe N, Maigre L, Santini S, Pinet E, Claverie JM et al. In vivo evolution of

463  

bacterial resistance in two cases of Enterobacter aerogenes infections during treatment with

464  

imipenem. PLoS One 2015;10:e0138828.

465  

59. Mecsas J, Rouviere PE, Erickson JW, Donohue TJ, Gross CA. The activity of

466  

sigma E, an Escherichia coli heat-inducible sigma-factor, is modulated by expression of outer

467  

membrane proteins. Genes Dev 1993;7:2618-2628.

468  

60. Missiakas D, Mayer MP, Lemaire M, Georgopoulos C, Raina S. Modulation of

469  

the Escherichia coli sigmaE (RpoE) heat-shock transcription-factor activity by the RseA,

470  

RseB and RseC proteins. Mol Microbiol. 1997;24(2):355-371.

471  

61. Rhodius VA, Suh WC, Nonaka G, West J, Gross CA. Conserved and variable

472  

functions of the sigmaE stress response in related genomes. PLoS Biol. 2006;4:e2.

473  

62. Noor R, Murata M, Nagamitsu H, Klein G, Raina S, Yamada M. Dissection of

474  

sigma(E)-dependent cell lysis in Escherichia coli: roles of RpoE regulators RseA, RseB and

475  

periplasmic folding catalyst PpiD. Genes Cells 2009;14:885-899.

476  

63. Gogol EB, Rhodius VA, Papenfort K, Vogel J, Gross CA. Small RNAs endow a

477  

transcriptional activator with essential repressor functions for single-tier control of a global

478  

stress regulon. Proc Natl Acad Sci U S A 2011;108:12875-12880.

479  

64. Klein G, Lindner B, Brade H, Raina S. Molecular basis of lipopolysaccharide

480  

heterogeneity in Escherichia coli: envelope stress-responsive regulators control the

481  

incorporation of glycoforms with a third 3-deoxy-α-D-manno-oct-2-ulosonic acid and

482  

rhamnose. J Biol Chem 2011;286:42787-42807.

483  

65. Klein G, Kobylak N, Lindner B, Stupak A, Raina S. Assembly of

484  

lipopolysaccharide in Escherichia coli requires the essential LapB heat shock protein. J Biol

485  

Chem 2014;289:14829-14853.

486  

66. Guo MS, Updegrove TB, Gogol EB, Shabalina SA, Gross CA, Storz G. MicL, a

487  

new σE-dependent sRNA, combats envelope stress by repressing synthesis of Lpp, the major

488  

outer membrane lipoprotein. Genes Dev 2014;28:1620-1634.

489  

67. Klein G, Stupak A, Biernacka D, Wojtkiewicz P, Lindner B

et al. Multiple 490  

transcriptional factors regulate transcription of the rpoE gene in Escherichia coli under

491  

different growth conditions and when the lipopolysaccharide biosynthesis is defective. J Biol

492  

Chem 2016;291:22999-23019.

493  

68. Ades SE. Regulation by destruction: design of the sigmaE envelope stress response.

494  

Curr Opin Microbiol 2008;11:535-540.

495  

(19)

69. Walsh NP, Alba BM, Bose B, Gross CA, Sauer RT. OMP peptide signals initiate

496  

the envelope-stress response by activating DegS protease via relief of inhibition mediated by

497  

its PDZ domain. Cell 2003;113:61-71.

498  

70. Lima S, Guo MS, Chaba R, Gross CA, Sauer RT. Dual molecular signals mediate

499  

the bacterial response to outer-membrane stress. Science 2013;340(6134):837-841.

500  

71. Mizuno T, Chou MY, Inouye M. A unique mechanism regulating gene expression:

501  

translational inhibition by a complementary RNA transcript (micRNA). Proc Natl Acad Sci U

502  

S A 1984;81:1966-1970.

503  

72. Delihas N. Discovery and characterization of the first non-coding RNA that regulates

504  

gene expression, micF RNA: A historical perspective. World J Biol Chem 2015;6:272-280.

505  

73. Inouye M. The first demonstration of RNA interference to inhibit mRNA function.

506  

Gene 2016;592(2):332-333.

507  

74. Andersen J, Delihas N. micF RNA binds to the 5' end of ompF mRNA and to a

508  

protein from Escherichia coli. Biochemistry 1990;29:9249-9256.

509  

75. Delihas N, Forst S. MicF: an antisense RNA gene involved in response of

510  

Escherichia coli to global stress factors. J Mol Biol 2001;313:1-12.

511  

76. Ramani N, Hedeshian M, Freundlich M. micF antisense RNA has a major role in

512  

osmoregulation of OmpF in Escherichia coli. J Bacteriol 1994;176:5005-5010.

513  

77. Chou JH, Greenberg JT, Demple B. Post-transcriptional repression of Escherichia

514  

coli OmpF protein in response to redox stress: positive control of the micF antisense RNA by

515  

the soxRS locus. J Bacteriol 1993;175:1026-1031.

516  

78. Chen S, Zhang A, Blyn LB, Storz G. MicC, a second small-RNA regulator of Omp

517  

protein expression in Escherichia coli. J Bacteriol 2004;186:6689-6697.

518  

79. Prilipov A, Phale PS, Koebnik R, Widmer C, Rosenbusch JP. Identification and

519  

characterization of two quiescent porin genes, nmpC and ompN, in Escherichia coli B

E

. J

520  

Bacteriol 1998;180:3388-3392.

521  

80. Dam S, Pagès JM, Masi M. Dual regulation of the small RNA MicC and the

522  

quiescent porin OmpN in response to antibiotic stress in Escherichia coli. Antibiotics (Basel)

523  

2017;6:pii: E33.

524  

81. Guillier M, Gottesman S. Remodelling of the Escherichia coli outer membrane by

525  

two small regulatory RNAs. Mol Microbiol 2006;59:231-47.

526  

82. Guillier M, Gottesman S. The 5' end of two redundant sRNAs is involved in the

527  

regulation of multiple targets, including their own regulator. Nucleic Acids Res

528  

2008;36:6781-6794.

529  

(20)

83. Rasmussen AA, Eriksen M, Gilany K, Udesen C, Franch T

et al. Regulation of 530  

ompA mRNA stability: the role of a small regulatory RNA in growth phase-dependent

531  

control. Mol Microbiol 2005;58:1421-1429.

532  

84. Johansen J, Rasmussen AA, Overgaard M, Valentin-Hansen P. Conserved small

533  

non-coding RNAs that belong to the sigmaE regulon: role in down-regulation of outer

534  

membrane proteins. J Mol Biol 2006;364:1-8.

535  

85. Skovierova H, Rowley G, Rezuchova B, Homerova D, Lewis

et al. Identification 536  

of the sigmaE regulon of Salmonella enterica serovar Typhimurium. Microbiology

537  

2006;152:1347-1359.

538  

86. Douchin V, Bohn C, Bouloc P. Down-regulation of porins by a small RNA bypasses

539  

the essentiality of the regulated intramembrane proteolysis protease RseP in Escherichia coli.

540  

J Biol Chem 2006;281:12253-12259.

541  

87. Papenfort K, Pfeiffer V, Mika F, Lucchini S, Hinton JC et al. SigmaE-dependent

542  

small RNAs of Salmonella respond to membrane stress by accelerating global omp mRNA

543  

decay. Mol Microbiol 2006 Dec;62(6):1674-88.

544  

88. Bossi L, Figueroa-Bossi N. A small RNA downregulates LamB maltoporin in

545  

Salmonella. Mol Microbiol 2007;65(3):799-810.

546  

89. Udekwu KI, Wagner EG. Sigma E controls biogenesis of the antisense RNA MicA.

547  

Nucleic Acids Res 2007;35:1279-1288.

548  

90. Johansen J, Eriksen M, Kallipolitis B, Valentin-Hansen P. Down-regulation of

549  

outer membrane proteins by noncoding RNAs: unraveling the cAMP-CRP- and sigmaE-

550  

dependent CyaR-OmpX regulatory case. J Mol Biol. 2008;383:1-9.

551  

91. Castillo-Keller M, Vuong P, Misra R. Novel mechanism of Escherichia coli porin

552  

regulation. J Bacteriol 2006;188:576-586.

553  

92. Doménech-Sánchez A, Hernández-Allés S, Martínez-Martínez L, Benedí VJ,

554  

Albertí S. Identification and characterization of a new porin gene of Klebsiella pneumoniae:

555  

its role in beta-lactam antibiotic resistance. J Bacteriol 1999;181:2726-2732.

556  

93. Knopp M, Andersson DI. Amelioration of the fitness costs of antibiotic resistance

557  

due to reduced outer membrane permeability by upregulation of alternative porins. Mol Biol

558  

Evol 2015;32:3252-3263.

559  

94. Pomposiello PJ, Bennik MH, Demple B. Genome-wide transcriptional profiling of

560  

the Escherichia coli responses to superoxide stress and sodium salicylate. J Bacteriol

561  

2001;183:3890-902.

562  

(21)

95. Bornet C, Davin-Régli A, Bosi C, Pagès JM, Bollet C. Imipenem resistance of

563  

Enterobacter aerogenes mediated by outer membrane permeability. J Clin Microbiol

564  

2000;38:1048-1052.

565  

96. Bornet C, Chollet R, Malléa M, Chevalier J, Davin-Régli A

et al.

Imipenem and

566  

expression of multidrug efflux pump in Enterobacter aerogenes. Biochem Biophys Res

567  

Commun 2003;301:985-990.

568  

97. Thiolas A, Bornet C, Davin-Régli A, Pagès JM, Bollet C. Resistance to imipenem,

569  

cefepime, and cefpirome associated with mutation in Omp36 osmoporin of Enterobacter

570  

aerogenes. Biochem Biophys Res Commun 2004;317(3):851-856.

571  

98. Thiolas A, Bollet C, La Scola B, Raoult D, Pagès JM. Successive emergence of

572  

Enterobacter aerogenes strains resistant to imipenem and colistin in a patient. Antimicrob

573  

Agents Chemother 2005;49:1354-1358.

574  

99. Lavigne JP, Sotto A, Nicolas-Chanoine MH, Bouziges N, Pagès JM

et al. An 575  

adaptive response of Enterobacter aerogenes to imipenem: regulation of porin balance in

576  

clinical isolates. Int J Antimicrob Agents 2013;41:130-136.

577  

100. Pavez M, Vieira C, de Araujo MR, Cerda A, de Almeida LM

et al.

Molecular

578  

mechanisms of membrane impermeability in clinical isolates of Enterobacteriaceae exposed

579  

to imipenem selective pressure. Int J Antimicrob Agents 2016;48:78-85.

580  

101. Cowan SW, Schirmer T, Rummel G, Steiert M, Ghosh R et al. Crystal structures explain 581  

functional properties of two E. coli porins. Nature 1992;358:727-733.

582  

102. Baslé A, Rummel G, Storici P, Rosenbusch JP, Schirmer T. Crystal structure of

583  

osmoporin OmpC from E. coli at 2.0 A. J Mol Biol 2006;362:933-942.

584  

103. Nikaido H, Rosenberg EY. Porin channels in Escherichia coli: studies with

585  

liposomes reconstituted from purified proteins. J Bacteriol 1983;153:241-252.

586  

104. Nikaido H, Rosenberg EY, Foulds, J. Porin channels in Escherichia coli: studies

587  

with beta-lactams in intact cells. J Bacteriol 1983;153:232-240.

588  

105. Kojima S, Nikaido H. High salt concentrations increase permeability through OmpC

589  

channels of Escherichia coli. J Biol Chem 2014;289:26464-26473.

590  

106. Dé E, Baslé A, Jaquinod M, Saint N, Malléa M

et al.

A new mechanism of

591  

antibiotic resistance in Enterobacteriaceae induced by a structural modification of the major

592  

porin. Mol Microbiol 2001;41:189-198.

593  

107. Simonet V, Malléa M, Pagès JM. Substitutions in the eyelet region disrupt cefepime

594  

diffusion through the Escherichia coli OmpF channel. Antimicrob Agents Chemother

595  

2000;44:311-315.

596  

(22)

108. Low AS, MacKenzie FM, Gould IM, Booth IR. Protected environments allow

597  

parallel evolution of a bacterial pathogen in a patient subjected to long-term antibiotic

598  

therapy. Mol Microbiol 2001;42:619-630.

599  

109. Lou H, Chen M, Black SS, Bushell SR, Ceccarelli M.

et al. Altered antibiotic 600  

transport in OmpC mutants isolated from a series of clinical strains of multi-drug resistant E.

601  

coli. PLoS One 2011;6(10):e25825.

602  

110. El-Mowafi SA, Alumasa JN, Ades SE, Keiler KC. Cell-based assay to identify

603  

inhibitors of the Hfq-sRNA regulatory pathway. Antimicrob Agents Chemother

604  

2014;58:5500-5509.

605  

111. Childs-Disney JL, Disney MD. Small molecule targeting of a microRNA associated

606  

with hepatocellular carcinoma. ACS Chem Biol 2016;11:375-380.

607  

(23)

Key figure: Major regulatory pathways of porin regulation in E. coli: EnvZ/OmpR [46],

608  

CpxAR and sigma E (σ

E

) [35] stress response systems are shown, along with known inducing

609  

cues and targets relevant to porin regulation. The upregulation is shown with thick green

610  

arrows, while the downregulation is shown with red lines. In the EnvZ/OmpR TCS,

611  

activation of the response regulator OmpR results in phosphorylated and OmpR∼P

612  

downregulates the expression of OmpF both at the transcriptional and post-transcriptional

613  

levels, the latter via the MicF sRNA. The mar-sox-rob regulons also downregulate OmpF

614  

expression via MicF. Both the CpxAR and

σE

responses are activated by a variety of

615  

envelope stresses. For clarity, only periplasmic misfolded OMPs are represented here. On one

616  

hand, CpxR

P alters expression of multiple genes, including that of micF. On the other hand,

617  

the anti-sigma factor RseA is degraded by the successive action of two proteases, DegS and

618  

RseP at the periplasmic and the cytoplasmic site. Another protease, ClpXP specifically

619  

degrades the cytoplasmic RseA portion bound to

σE

, leading to its release. A number of σ

E

-

620  

regulated sRNAs are indicated: MicC [78] downregulates OmpC and is coupled with ompN

621  

upregulation [80]; sRNA regulation of porins via CyaR [90], IpeX [91], RseX [86] and RybB

622  

[84, 88] are shown accompanied by their activators and porin targets; CyaR negatively

623  

regulates the expression of single channeled porin OmpX [30], which in turn negatively

624  

regulates the major porin OmpC. The details of all these interconnected pathways are

625  

discussed thoroughly in the text.

626   627  

(24)

  628  

Références

Documents relatifs

Wild type and complemented phoPQEcl mutant strains grown in high Mg2+ demonstrated some susceptibility to 5 and 10 g/ml of colistin Fig 5A, High Mg2+, suggesting

Taking into account the direction of the local magnetic field the flow direction of the protons implies that they are coming from the expected reconnection site (magnetopause close

Comme Jung, écrivons notre livre rouge Un moment de sa vie plongé dans la détresse Carl Gustav Jung extirpe ses faiblesses. Sa rupture avec Freud crée le questionnement Rejetant

Deletion of the acrB gene from strain KPBj1 Rev, devoid of the rarA gene and the oqxRAB operon, and normally producing the AcrAB pump (Table S1), resulted in nalidixic acid

(6) to support Member States to mobilize adequate predictable and sustained funding and human and financial resources and investment through national, bilateral

The draft Framework for Accelerating Action to Fight Antimicrobial Resistance in the Western Pacific Region reiterates the need for the health sector to work beyond health,

This manual for developing national action plans to address antimicrobial resistance has been developed at the request of the World Health Assembly to assist countries in the

Three countries had no guidelines for surveillance of use/or sales of antimicrobials (phase 1); in four countries, the national policy and plan on surveillance of